EP1345946B1 - Makroheterocyclische verbindungen als kinase inhibitoren - Google Patents

Makroheterocyclische verbindungen als kinase inhibitoren Download PDF

Info

Publication number
EP1345946B1
EP1345946B1 EP01996227A EP01996227A EP1345946B1 EP 1345946 B1 EP1345946 B1 EP 1345946B1 EP 01996227 A EP01996227 A EP 01996227A EP 01996227 A EP01996227 A EP 01996227A EP 1345946 B1 EP1345946 B1 EP 1345946B1
Authority
EP
European Patent Office
Prior art keywords
compound
mmol
kinase
mixture
disorders
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
EP01996227A
Other languages
English (en)
French (fr)
Other versions
EP1345946A1 (de
Inventor
Gee-Hong Kuo
Catherine Prouty
Alan Deangelis
Han-Cheng Zhang
Peter. Conolly
William.V. Murray
Chandra R. Shah
Bruce E. Maryanoff
Kimberly B. White
Lan Shen
Bruce. Conway
Keith. Demarest
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Janssen Pharmaceuticals Inc
Original Assignee
Ortho McNeil Pharmaceutical Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ortho McNeil Pharmaceutical Inc filed Critical Ortho McNeil Pharmaceutical Inc
Publication of EP1345946A1 publication Critical patent/EP1345946A1/de
Application granted granted Critical
Publication of EP1345946B1 publication Critical patent/EP1345946B1/de
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/22Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed systems contains four or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/22Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains four or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/22Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains four or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D515/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D515/22Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains four or more hetero rings

Definitions

  • This invention is directed to certain novel macroheterocyclic compounds, methods for producing such compounds and methods for treating or ameliorating a kinase or dual-kinase mediated disorder. More particularly, this invention is directed to macroheterocyclic 1 H -indole, 1 H -pyrrolo[2,3- b ]pyridine, 1 H -pyrazolo[3,4- b ]pyridine, and 1 H -indazole compounds useful as selective kinase or dual-kinase inhibitors, methods for producing such compounds and methods for treating or ameliorating a kinase or dual-kinase mediated disorder.
  • It is an object of the present invention to provide macroheterocyclic 1 H -indole, 1 H -pyrrolo[2,3- b ]pyridine, 1 H -pyrazolo[3,4- b ]pyridine, and 1 H -indazole compounds useful as a kinase or dual-kinase inhibitor i.e., a compound capable of inhibiting two or more kinases such as, for example, a kinase selected from protein kinase C or glycogen synthase kinase-3; and, more particularly, a kinase selected from protein kinase C ⁇ , protein kinase C ⁇ -II, protein kinase C ⁇ or glycogen synthase kinase-3 ⁇ ), methods for their production and methods for treating or ameliorating a kinase or dual-kinase mediated disorder.
  • a kinase or dual-kinase inhibitor i
  • the present invention provides a macroheterocyclic compound of Formula (Ia1): wherein R 4 , R 2 and R 5 are dependently selected from: R 4 R 2 R 5 -(CB 2 ) 2 - -O-(CH 2 ) 2 -O- -(CH 2 ) 2 -; -(CB 2 ) 2 - -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O- -(CH 2 ) 2 -; -(CH 2 ) 2 - -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -; -(CH 2 ) 2 - -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -; -(CH 2 ) 2 - -O-(CH 2 ) 2 -O
  • the invention also provides a compound of Formula (Ibl): wherein R 4 , R 2 and R 5 are dependently selected from: R 4 R 2 R 5 -(CH 2 ) 2 - -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O- -(CH 2 ) 2 -; -(CH 2 ) 2 - -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O- -(CH 2 ) 2 -; -(CH 2 ) 2 - -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -; -(CH 2 ) 2 - -O-(CH 2 ) 2 -N(Et)-(CH 2 ) 2 -O- -(CH 2 ) 2 -; -(CH
  • the invention further provides a compound of Formula (Ifl): wherein R 4 , R 2 and R 5 are dependently selected from: R 4 R 2 R 5 -(CH 2 ) 2 - -O-(CH 2 ) 2 -N(Me)-(CH 2 ) 2 -O- -(CH 2 ) 2 -; -(CH 2 ) 2 - -O-(CH 2 ) 2 -N(Et)-(CH 2 ) 2 -O- -(CH 2 ) 2 -; and, -(CH 2 ) 2 - -O-(CH 2 ) 2 -N(2-OMe-Et)-(CH 2 ) 2 -O- -(CH 2 ) 2 -. and pharmaceutically acceptable salts thereof.
  • the invention also provides a compound of Formula (Ii1): wherein R 4 , R 2 and R 5 are dependently selected from: R 4 R 2 R 5 -CH 2 - -1,3-phenyl- -CH 2 -; and, -CH 2 - -2,6-pyridinyl- -CH 2 -. and pharmaceutically acceptable salts thereof.
  • the invention also provides a compound of Formula (Ijl): wherein R 4 , R 2 and R 5 are dependently selected from: R 4 R 2 R 5 -(CH 2 ) 2 - -O-(CH 2 ) 2 -O- -(CH 2 ) 2 - ; and -(CH 2 ) 2 - -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O- -(CH 2 ) 2 - and pharmaceutically acceptable salts thereof.
  • the present invention is directed to macroheterocyclic compounds useful as a selective kinase or dual-kinase inhibitor, preferably as inhibitors of kinases selected from protein kinase C or glycogen synthase kinase-3; and, more particularly, a kinase selected from protein kinase C ⁇ , protein kinase C ⁇ -II, protein kinase C ⁇ or glycogen synthase kinase-3 ⁇ .
  • the present invention is also directed to methods for producing the instant macroheterocyclic compounds and pharmaceutical compositions and medicaments thereof.
  • the present invention is further directed to methods for treating or ameliorating a kinase or dual-kinase mediated disorder.
  • the method of the present invention is directed to treating or ameliorating a kinase mediated disorder such as, but not limited to, cardiovascular diseases, diabetes, diabetes-associated disorders, inflammatory diseases, immunological disorders, dermatological disorders, oncological disorders and CNS (Central Nervous System) disorders.
  • cardiovascular diseases such as, but not limited to, cardiovascular diseases, diabetes, diabetes-associated disorders, inflammatory diseases, immunological disorders, dermatological disorders, oncological disorders and CNS (Central Nervous System) disorders.
  • CNS Central Nervous System
  • Exemplified compounds of the present invention include a compound of Formula (Ia) selected from a compound of Formula (Ia1): wherein R 4 , R 2 and R 5 are dependently selected from: Cpd R 4 R 2 R 5 4 -(CH 2 ) 2 - -O-(CH 2 ) 2 -O- -(CH 2 ) 2 - 5 -(CH 2 ) 2 - -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O- -(CH 2 ) 2 - 6 -(CH 2 ) 2 - -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O- -(CH 2 ) 2 - 7 -(CH 2 ) 2 - -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2
  • Exemplified compounds of the present invention include a compound of Formula (Ib) selected from a compound of Formula (Ib1): wherein R 4 , R 2 and R 5 are dependently selected from: Cpd R 4 R 2 R 5 1 -(CH 2 ) 2 - -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O- -(CH 2 ) 2 - 2 -(CH 2 ) 2 - -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O- -(CH 2 ) 2 - 3 -(CH 2 ) 2 - -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 - 18 -(CH 2 ) 2 - -O-(CH 2 ) 2 -
  • Exemplified compounds of the present invention include a compound of Formula (If) selected from a compound of Formula (Ifl): wherein R 4 , R 2 and R 5 are dependently selected from: Cpd R 4 R 2 R 5 16 -(CH 2 ) 2 - -O-(CH 2 ) 2 -N(Me)-(CH 2 ) 2 -O- -(CH 2 ) 2 - 17 -(CH 2 ) 2 - -O-(CH 2 ) 2 -N(Et)-(CH 2 ) 2 -O- -(CH 2 ) 2 - 29 -(CH 2 ) 2 - -O-(CH 2 ) 2 -N(2-OMe-Et)-(CH 2 ) 2 -O- -(CH 2 ) 2 -
  • Exemplified compounds of the present invention include a compound of Formula (Ii) selected from a compound of Formula (Ii1): wherein R 4 , R 2 and R 5 are dependently selected from: Cpd R 4 R 2 R 5 8 -CH 2 - -1,3-phenyl- -CH 2 - 9 -CH 2 - -2,6-pyridinyl- -CH 2 -
  • Exemplified compounds of the present invention include a compound of Formula (Ij) selected from a compound of Formula (Ij1): wherein R 4 , R 2 and R 5 are dependently selected from: Cpd R 4 R 2 R 5 10 -(CH 2 ) 2 - -O-(CH 2 ) 2 -O- -(CH 2 ) 2 - 11 -(CH 2 ) 2 - -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O-(CH 2 ) 2 -O- -(CH 2 ) 2 -
  • the compounds of the present invention may also be present in the form of pharmaceutically acceptable salts.
  • the salts of the compounds of this invention refer to non-toxic "pharmaceutically acceptable salts" (Ref. International J. Pharm., 1986, 33 , 201-217; J. Pharm.Sci., 1997 (Jan), 66, 1, 1).
  • Other salts may, however, be useful in the preparation of compounds according to this invention or of their pharmaceutically acceptable salts.
  • organic or inorganic acids include, but are not limited to, hydrochloric, hydrobromic, hydriodic, perchloric, sulfuric, nitric, phosphoric, acetic, propionic, glycolic, lactic, succinic, maleic, fumaric, malic, tartaric, citric, benzoic, mandelic, methanesulfonic, hydroxyethanesulfonic, benezenesulfonic, oxalic, pamoic, 2-naphthalenesulfonic, p-toluenesulfonic, cyclohexanesulfamic, salicylic, saccharinic or trifluoroacetic acid.
  • Organic or inorganic bases include, but are not limited to, basic or cationic salts such as benzathine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine, procaine, aluminum, calcium, lithium, magnesium, potassium, sodium and zinc.
  • basic or cationic salts such as benzathine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine, procaine, aluminum, calcium, lithium, magnesium, potassium, sodium and zinc.
  • the present invention includes within its scope prodrugs of the compounds of this invention.
  • prodrugs will be functional derivatives of the compounds, which are readily convertible in vivo into the required compound.
  • the term “administering” shall encompass the treatment of the various disorders described with the compound specifically disclosed or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the subject.
  • Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in " Design of Prodrugs ", ed. H. Bundgaard, Elsevier, 1985.
  • the compounds according to this invention may accordingly exist as enantiomers. Where the compounds possess two or more chiral centers, they may additionally exist as diastereomers. Where the processes for the preparation of the compounds according to the invention give rise to mixture of stereoisomers, these isomers may be separated by conventional techniques such as. preparative chromatography.
  • the compounds may be prepared in racemic form or individual enantiomers may be prepared by standard techniques known to those skilled in the art, for example, by enantiospecific synthesis or resolution, formation of diastereomeric pairs by salt formation with an optically active acid, followed by fractional crystallization and regeneration of the free base.
  • the compounds may also be resolved by formation of diastereomeric esters or amides, followed by chromatographic separation and removal of the chiral auxiliary. Alternatively, the compounds may be resolved using a chiral HPLC column. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present invention.
  • alkyl refers to a saturated straight or branched chain consisting solely of 1-8 hydrogen substituted carbon atoms; preferably, 1-6 hydrogen substituted carbon atoms; and, most preferably, 1-4 hydrogen substituted carbon atoms.
  • alkenyl refers to a partially unsaturated straight or branched alkyl chain that contains at least one double bond.
  • alkynyl refers to a partially unsaturated straight or branched alkyl chain that contains at least one triple bond.
  • alkoxy refers to -O-alkyl, where alkyl is as defined supra.
  • alkylthio refers to -S-alkyl, where alkyl is as defined supra.
  • a carboxyl group is a carbonyl with a terminal OH group.
  • the branched alkyl chain may be substituted on the linking alkyl chain, the branch of the linking alkyl chain or on both.
  • cycloalkyl refers to a saturated or partially unsaturated cyclic alkyl ring consisting of 3-8 hydrogen substituted carbon atoms. Examples include, and are not limited to, cyclopropyl, cyclopentyl, cyclohexyl or cycloheptyl.
  • spirocycloalkyl refers to a cycloalkyl ring sharing a single ring carbon with another attached ring.
  • heterocyclyl refers to a saturated or partially unsaturated ring having five members of which at least one member is a N, O or S atom and which optionally contains one additional O atom or one, two or three additional N atoms, a saturated or partially unsaturated ring having six members of which one, two or three members are a N atom, a saturated or partially unsaturated bicyclic ring having nine members of which at least one member is a N, O or S atom and which optionally contains one, two or three additional N atoms and a saturated or partially unsaturated bicyclic ring having ten members of which one, two or three members are a N atom.
  • Examples include, and are not limited to, pyrrolinyl, pyrrolidinyl, dioxolanyl, imidazolinyl, imidazolidinyl, pyrazolinyl, pyrazolidinyl, piperidinyl, morpholinyl or piperazinyl.
  • spiroheterocyclyl refers to a heterocyclyl ring sharing a single ring carbon with another attached ring.
  • aryl refers to an aromatic monocyclic ring system containing 5-6 hydrogen substituted carbon atoms or an aromatic bicyclic ring system containing 9-14 hydrogen substituted carbon atoms. Examples include, and are not limited to, phenyl, naphthalenyl or anthracenyl.
  • heteroaryl refers to an aromatic monocyclic ring system containing five members of which at least one member is a N, O or S atom and which optionally contains one, two or three additional N atoms, an aromatic monocyclic ring having six members of which one, two or three members are a N atom, an aromatic bicyclic ring having nine members of which at least one member is a N, O or S atom and which optionally contains one, two or three additional N atoms and an aromatic bicyclic ring having ten members of which one, two or three members are a N atom.
  • Examples include, and are not limited to, furyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, quinolinyl or isoquinolinyl.
  • halo or halogen refers to a fluoro, chloro, bromo or iodo atom.
  • An embodiment of the invention is a pharmaceutical composition comprising a pharmaceutically acceptable carrier and any of the compounds described above.
  • Illustrative of the invention is a pharmaceutical composition made by mixing any of the compounds described above and a pharmaceutically acceptable carrier.
  • Another illustration of the invention is a process for making a pharmaceutical composition comprising mixing any of the compounds described above and a pharmaceutically acceptable carrier.
  • Further illustrative of the present invention are pharmaceutical compositions comprising one or more compounds of this invention in association with a pharmaceutically acceptable carrier.
  • composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combinations of the specified ingredients in the specified amounts.
  • the compounds of the present invention are selective kinase or dual-kinase inhibitors useful in a method for treating or ameliorating a kinase or dual-kinase mediated disorder.
  • the kinase is selected from protein kinase C or glycogen synthase kinase-3 and more preferably, the kinase is selected from protein kinase C ⁇ , protein kinase C ⁇ -II, protein kinase C ⁇ or glycogen synthase kinase-3 ⁇ .
  • the compounds of this invention demonstrate inhibitory activity for a number of other kinases as well.
  • PKC Protein kinase C
  • the PKC family is composed of twelve isoforms that are further classified into 3 subfamilies: the calcium dependent classical PKC isoforms alpha ( ⁇ ), beta-I ( ⁇ -I), beta-II ( ⁇ -II) and gamma ( ⁇ ); the calcium independent PKC isoforms delta ( ⁇ ), epsilon ( ⁇ ), eta ( ⁇ ), theta ( ⁇ ) and mu ( ⁇ ); and, the atypical PKC isoforms zeta ( ⁇ ), lambda ( ⁇ ) and iota ( ⁇ ).
  • PKC vascular endothelial growth factor
  • VEGF vascular endothelial growth factor
  • a diabetes-linked elevation of the ⁇ isoform in human platelets has been correlated with the altered response of the platelets to agonists (Bastyr III, E. J. and Lu, J., Diabetes, 1993, 42, (Suppl. 1) 97A).
  • the human vitamin D receptor has been shown to be selectively phosphorylated by PKC ⁇ . This phosphorylation has been linked to alterations in the functioning of the receptor (Hsieh, et al., Proc. Natl.
  • PKC activity plays an important role in cardiovascular diseases. Increased PKC activity in the vasculature has been shown to cause increased vasoconstriction and hypertension (Bilder, G. E., et al., J. Pharmacol. Exp. Ther., 1990, 252, 526-530). PKC inhibitors block agonist-induced smooth muscle cell proliferation (Matsumoto, H. and Sasaki, Y., Biochem. Biophys. Res. Commun ., 1989, 158, 105-109).
  • PKC ⁇ triggers events leading to the induction of Egr-1 (Early Growth Factor-1) and tissue factor under hypoxic conditions (as part of the oxygen deprivation-mediated pathway for triggering procoagulant events) (Yan, S-F, et al., J. Biol. Chem., 2000, 275, 16, 11921-11928).
  • Egr-1 Early Growth Factor-1
  • tissue factor under hypoxic conditions as part of the oxygen deprivation-mediated pathway for triggering procoagulant events
  • PAI-1 Plasminogen Activator Inhibitor-1
  • PKC inhibitors are useful in treating cardiovascular ischemia and improving cardiac function following ischemia (Muid, R. E., et al., FEBS Lett ., 1990, 293, 169-172; Sonoki,, H. et al., Kokyu-To Junkan, 1989, 37 , 669-674). Elevated PKC levels have been correlated with an increase in platelet function in response to agonists (Bastyr III, E. J. and Lu, J., Diabetes , 1993, 42 , (Suppl. 1) 97A). PKC has been implicated in the biochemical pathway in the platelet-activating factor (PAF) modulation of microvascular permeability (Kobayashi, et al., Amer.
  • PAF platelet-activating factor
  • PKC inhibitors affect agonist-induced aggregation in platelets (Toullec, D., et al., J. Biol. Chem ., 1991 , 266 , 15771-15781). Accordingly, PKC inhibitors may be indicated for use in treating cardiovascular disease, ischemia, thrombotic conditions, atherosclerosis and restenosis.
  • PKC- ⁇ -II activation of the PKC- ⁇ -II isoform plays an important role in diabetic vascular complications such as retinopathy (Ishii, H., et al., Science, 1996, 272, 728-731) and PKC ⁇ has been implicated in development of the cardiac hypertrophy associated with heart failure (X. Gu, et al., Circ. Res., 1994, 75, 926; R. H. Strasser, et al., Circulation, 1996, 94 , 1551).
  • Overexpression of cardiac PKC ⁇ II in transgenic mice caused cardiomyopathy involving hypertrophy, fibrosis and decreased left ventricular function (H. Wakasaki, et al., Proc. Natl. Acad. Sci. USA, 1997, 94 , 9320).
  • PKC inhibitors block inflammatory responses such as the neutrophil oxidative burst, CD3 down-regulation in T-lymphocytes and phorbol-induced paw edema (Twoemy, B., et al., Biochem. Biophys. Res. Commun ., 1990 , 171, 1087-1092; Mulqueen, M. J., et al. Agents Actions, 1992, 37, 85-89).
  • PKC ⁇ has an essential role in the degranulation of bone marrow-derived mast cells, thus affecting cell capacity to produce IL-6 (Interleukin-6) (Nechushtan, H., et al., Blood, 2000 (March), 95, 5, 1752-1757).
  • PKC plays a role in enhanced ASM (Airway Smooth Muscle) cell growth in rat models of two potential risks for asthma: hyperresponsiveness to contractile agonists and to growth stimuli (Ren, S, et al., Am. J. Physiol., 2000 , 278, (4, Pt. 1), E656-E662).
  • ASM Airway Smooth Muscle
  • PKC ⁇ -1 overexpression augments an increase in endothelial permeability, suggesting an important function in the regulation of the endothelial barrier (Nagpala, P.G., et al., J. Cell Physiol., 1996 , 2 , 249-55).
  • PKC ⁇ mediates activation of neutrophil NADPH oxidase by PMA and by stimulation of Fc ⁇ receptors in neutrophils (Dekker, L.V., et al., Biochem. J., 2000 , 347 , 285-289).
  • PKC inhibitors may be indicated for use in treating inflammation and asthma.
  • PKC may be useful in treating or ameliorating certain immunological disorders. While one study suggests that HCMV (Human Cytomegalovirus) inhibition is not correlated with PKC inhibition (Slater, M.J., et al., Biorg. & Med. Chem ., 1999, 7, 1067-1074), another study showed that the PKC signal transduction pathway synergistically interacted with the cAMP-dependent PKA pathway to activate or increase HIV-1 transcription and viral replication and was abrogated with a PKC inhibitor (Rabbi, M.F., et al., Virology , 1998 (June 5), 245 , 2, 257-69). Therefore, an immunological disorder may be treated or ameliorated as a function of the affected underlying pathway's response to up- or down-regulation of PKC.
  • HCMV Human Cytomegalovirus
  • PKC ⁇ deficiency also results in an immunodeficiency characterized by impaired humoral immune responses and a reduced B cell response, similar to X-linked immunodeficiency in mice and plays an important role in antigen receptor-mediated signal transduction (Leitges, M., et al., Science (Wash., D.C.), 1996, 273 , 5276, 788-789). Accordingly, transplant tissue rejection may be ameliorated or prevented by suppressing the immune response using a PKC ⁇ inhibitor.
  • Abnormal activity of PKC has been linked to dermatological disorders characterized by abnormal proliferation of keratinocytes, such as psoriasis (Horn, F., et al., J. Invest. Dermatol., 1987 , 88 , 220-222; Raynaud, F. and Evain-Brion, D., Br. J. Dermatol ., 1991, 124, 542-546).
  • PKC inhibitors have been shown to inhibit keratinocyte proliferation in a dose-dependent manner (Hegemann, L., et al., Arch. Dermatol. Res., 1991 , 283, 456-460 ; Bollag, W. B., et al., J. Invest. Dermatol., 1993 , 100 , 240-246).
  • PKC activity has been associated with cell growth, tumor promotion, uncontrolled cell growth and cancer (Rotenberg, S. A. and Weinstein, 1. B., Biochem. Mol. Aspects Sel. Cancer,1991, 1 , 25-73; Ahmad, et al., Molecular Pharmacology, 1993, 43 , 858-862); PKC inhibitors are known to be effective in preventing tumor growth in animals (Meyer, T., et al., Int. J. Cancer, 1989 , 43 , 851-856; Akinagaka, S., et al., Cancer Res., 1991, 51 , 4888-4892).
  • PKC ⁇ -1 and ⁇ -2 expression in differentiated HD3 colon carcinoma cells blocked their differentiation, enabling them to proliferate in response to basic FGF (Fibroblast Growth Factor) like undifferentiated cells, increasing their growth rate and activating several MBP (Myelin-Basic Protein) kinases, including p57 MAP (Mitogen-Activated Protein) kinase (Sauma, S., et al., Cell Growth Differ., 1996 , 7 , 5, 587-94).
  • PKC ⁇ inhibitors having an additive therapeutic effect in combination with other anti-cancer agents, inhibited the growth of lymphocytic leukemia cells (Konig, A., et al., Blood, 1997 , 90, 10, Suppl.
  • PKC inhibitors enhanced MMC (Mitomycin-C) induced apoptosis in a time-dependent fashion in a gastric cancer cell-line, potentially indicating use as agents for chemotherapy-induced apoptosis (Danso, D., et al., Proc. Am. Assoc. Cancer Res., 1997 , 38 , 88 Meet., 92). Therefore, PKC inhibitors may be indicated for use in ameliorating cell and tumor growth, in treating or ameliorating cancers (such as leukemia or colon cancer) and as adjuncts to chemotherapy.
  • MMC Mitomycin-C
  • cancers such as leukemia or colon cancer
  • PKC ⁇ (by enhancing cell migration) may mediate some proangiogenic effects of PKC activation while PKC ⁇ may direct antiangiogenic effects of overall PKC activation (by inhibiting cell growth and proliferation) in capillary endothelial cells, thus regulating endothelial proliferation and angiogenesis (Harrington, E.O., et al., J. Biol. Chem., 1997, 272 , 11, 7390-7397).
  • PKC inhibitors inhibit cell growth and induce apoptosis in human glioblastoma cell lines, inhibit the growth of human astrocytoma xenografts and act as radiation sensitizers in glioblastoma cell lines (Begemann, M., et al., Anticancer Res. (Greece), 1998 (Jul-Aug), 18, 4A, 2275-82).
  • PKC inhibitors, in combination with other anti-cancer agents are radiation and chemosensitizers useful in cancer therapy (Teicher, B.A., et al., Proc. Am. Assoc. Cancer Res., 1998 , 39 , 89 Meet., 384).
  • PKC ⁇ inhibitors by blocking the MAP kinase signal transduction pathways for VEGF (Vascular Endothelial Growth Factor) and bFGF (basic Fibrinogen Growth Factor) in endothelial cells, in a combination regimen with other anti-cancer agents, have an anti-angiogenic and antitumor effect in a human T98G glioblastoma multiforme xenograft model (Teicher, B.A., et al., Clinical Cancer Research, 2001 (March), 7, 634-640). Accordingly, PKC inhibitors may be indicated for use in ameliorating angiogenesis and in treating or ameliorating cancers (such as breast, brain, kidney, bladder, ovarian or colon cancers) and as adjuncts to chemotherapy and radiation therapy.
  • cancers such as breast, brain, kidney, bladder, ovarian or colon cancers
  • PKC activity plays a central role in the functioning of the CNS (Huang, K. P., Trends Neurosci., 1989, 12 , 425-432) and PKC is implicated in Alzheimer's disease (Shimohama, S., et al., Neurology, 1993 , 43 , 1407-1413) and inhibitors have been shown to prevent the damage seen in focal and central ischemic brain injury and brain edema (Hara, H., et al., J. Cereb. Blood Flow Metab., 1990, 10, 646-653; Shibata, S., et al., Brain Res., 1992, 594, 290-294). Accordingly, PKC inhibitors may be indicated for use in treating Alzheimers disease and in treating neurotraumatic and ischemia-related diseases.
  • PKC ⁇ as a component of the phosphoinositide 2 nd messenger system
  • muscarinic acetylcholine receptor expression in an amygdala-kindled rat model has been associated with epilepsy, serving as a basis for the rat's permanent state of hyperexcitability (Beldhuis, H.J.A., et al., Neuroscience, 1993 , 55 , 4, 965-73). Therefore, PKC inhibitors may be indicated for use in treating epilepsy.
  • PKC has demonstrated a role in the pathology of conditions such as, but not limited to, cardiovascular diseases, diabetes, diabetes-associated disorders, inflammatory diseases, immunological disorders, dermatological disorders, oncological disorders and central nervous system disorders.
  • Glycogen synthase kinase-3 (GSK-3) is a serine/threonine protein kinase composed of two isoforms ( ⁇ and ⁇ ) which are encoded by distinct genes.
  • GSK-3 is one of several protein kinases which phosphorylate glycogen synthase (GS) (Embi, et al., Eur. J. Biochem , 1980 , 107 , 519-527).
  • the ⁇ and ⁇ isoforms have a monomeric structure of 49 and 47kD respectively and are both found in mammalian cells.
  • Type II diabetes or Non-Insulin Dependent Diabetes Mellitus, NIDDM
  • NIDDM Non-Insulin Dependent Diabetes Mellitus
  • Hyperglycemia is due to insulin resistance in the liver, muscle and other tissues coupled with inadequate or defective secretion of insulin from pancreatic islets.
  • Skeletal muscle is the major site for insulin-stimulated glucose uptake. In this tissue, glucose removed from the circulation is either metabolised through glycolysis and the TCA (tricarboxylic acid) cycle or stored as glycogen.
  • Muscle glycogen deposition plays the more important role in glucose homeostasis and Type II diabetic subjects have defective muscle glycogen storage.
  • the stimulation of glycogen synthesis by insulin in skeletal muscle results from the dephosphorylation and activation of glycogen synthase (Villar-Palasi C.
  • GSK-3 is responsible for phosphorylation and deactivation of GS, while glycogen bound protein phosphatase 1 (PP1G) dephosphorylates and activates GS. Insulin both inactivates GSK-3 and activates PP1G (Srivastava A.K. and Pandey S.K., Mol. and Cellular Biochem., 1998 , 182 , 135-141).
  • GSK-3 ⁇ and constitutively active GSK-3 ⁇ (S9A, S9e) mutants in HEK-293 cells resulted in suppression of glycogen synthase activity (Eldar-Finkelman, et al., PNAS, 1996, 93 , 10228-10233) and overexpression of GSK-3 ⁇ in CHO cells, expressing both insulin receptor and insulin receptor substrate 1 (IRS-1) resulted in impairment of insulin action (Eldar-Finkelman and Krebs, PNAS, 1997, 94 , 9660-9664).
  • GSK-3 ⁇ knockout mouse Studies on fibroblasts from the GSK-3 ⁇ knockout mouse indicate that inhibition of GSK-3 may be useful in treating inflammatory disorders or diseases through the negative regulation of NFkB activity (Hoeflich K. P., et al., Nature , 2000, 406, 86-90).
  • GSK-3 In addition to modulation of glycogen synthase activity, GSK-3 also plays an important role in the CNS disorders. GSK-3 inhibitors may be of value as neuroprotectants in the treatment of acute stroke and other neurotraumatic injuries (Pap and Cooper, J. Biol. Chem ., 1998, 273 , 19929-19932). Lithium, a low mM inhibitor of GSK-3, has been shown to protect cerebellar granule neurons from death (D'Mello, et al., Exp. Cell Res., 1994, 211, 332-338) and chronic lithium treatment has demonstrable efficacy in the middle cerebral artery occlusion model of stroke in rodents (Nonaka and Chuang, Neuroreport , 1998, 9(9), 2081-2084).
  • Tau and ⁇ -catenin, two known in vivo substrates of GSK-3, are of direct relevance in consideration of further aspects of the value of GSK-3 inhibitors in relation to treatment of chronic neurodegenerative conditions.
  • Tau hyperphosphorylation is an early event in neurodegenerative conditions such as Alzheimer's disease and is postulated to promote microtubule disassembly. Lithium has been reported to reduce the phosphorylation of tau, enhance the binding of tau to microtubules and promote microtubule assembly through direct and reversible inhibition of GSK-3 (Hong M. et al J. Biol. Chem ., 1997 , 272(40) , 25326-32).
  • ⁇ -catenin is phosphorylated by GSK-3 as part of a tripartite axin protein complex resulting in ⁇ -catenin degradation (Ikeda, et al., EMBO J ., 1998 , 17 , 1371-1384). Inhibition of GSK-3 activity is involved in the stabilization of catenin and promotes ⁇ -catenin-LEF-1/TCF transcriptional activity (Eastman, Grosschedl, Curr. Opin. Cell Biol., 1999, 11 , 233). Studies have also suggested that GSK-3 inhibitors may also be of value in the treatment of schizophrenia (Cotter D., et al.
  • GSK-3 inhibitors could have further therapeutic utility in the treatment of diabetes, inflammatory diseases, dermatological disorders and central nervous system disorders.
  • Embodiments of the present invention include the use of a compound of claims 1 to 5 for the preparation of a medicament for treating or ameliorating a kinase or dual-kinase mediated disorder in a subject in need thereof wherein a preferred method step comprises administering the kinase or dual-kinase inhibitor to a patient.
  • the therapeutically effective amount of the compounds of claims 1 to 5 exemplified in such a method is from about 0.001 mg/kg/day to about 300 mg/kg/day.
  • An individual compound of the present invention or a pharmaceutical composition thereof can be administered separately at different times during the course of therapy or concurrently in divided or single combination forms.
  • the term "administering" is therefore to be understood as embracing all such regimes of simultaneous or alternating treatment.
  • a compound or pharmaceutical composition thereof may advantageously be co-administered in combination with other agents for treating, reducing or ameliorating the effects of a kinase or dual-kinase mediated disorder.
  • a compound of claim 1 to 5 or pharmaceutical composition thereof may be used in combination with other agents, especially insulin or antidiabetic agents including, but not limited to, insulin secretagogues (such as sulphonylureas), insulin sensitizers including, but not limited to, glitazone insulin sensitizers (such as thiazolidinediones) or biguanides or a glucosidase inhibitors.
  • the combination product is a product that comprises the co-administration of a compound of claim 1 to 5 or a pharmaceutical composition thereof and an additional agent for treating or ameliorating a kinase or dual-kinase mediated disorder
  • the term combination product further comprises a product that is sequentially administered where the product comprises a compound of claim 1 to 5 or pharmaceutical composition thereof and an additional agent for treating or ameliorating a kinase or dual-kinase mediated disorder, administration of a pharmaceutical composition containing a compound of claim 1 to 5 or pharmaceutical composition thereof and an additional agent for treating or ameliorating a kinase or dual-kinase mediated disorder or the essentially simultaneous administration of a separate pharmaceutical composition containing a compound of claim 1 to 5 or pharmaceutical composition thereof and a separate pharmaceutical composition containing an additional agent for treating or ameliorating a kinase or dual-kinase mediated disorder.
  • subject refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
  • terapéuticaally effective amount means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue system, animal or human, that is being sought by a researcher, veterinarian, medical doctor, or other clinician, which includes alleviation of the symptoms of the disease or disorder being treated.
  • a particular compound of claim 1 to 5 is selected where it is therapeutically effective for a particular kinase or dual-kinase mediated disorder based on the modulation of the disorder through the demonstration of selective kinase or dual-kinase inhibition in response to that compound.
  • Experiments exemplifying selective kinase or dual-kinase inhibition are provided in the examples.
  • the usefulness of a compound of claim 1 to 5 as a selective kinase or dual-kinase inhibitor can be determined according to the methods disclosed herein and based on the data obtained to date, it is anticipated that a particular compound will be useful in inhibiting one or more kinase or dual-kinase mediated disorders and therefore is uesfull in one or more kinase or dual-kinase mediated disorders.
  • kinase or dual-kinase mediated disorders includes, and is not limited to, cardiovascular diseases, diabetes, diabetes-associated disorders, inflammatory diseases, immunological disorders, dermatological disorders, oncological disorders and CNS disorders.
  • Cardiovascular diseases include, and are not limited to, acute stroke, heart failure, cardiovascular ischemia, thrombosis, atherosclerosis, hypertension, restenosis, retinopathy of prematurity or age-related macular degeneration.
  • Diabetes includes insulin dependent diabetes or Type II non-insulin dependent diabetes mellitus.
  • Diabetes-associated disorders include, and are not limited to, impaired glucose tolerance, diabetic retinopathy, proliferative retinopathy, retinal vein occlusion, macular edema, cardiomyopathy, nephropathy or neuropathy.
  • Inflammatory diseases include, and are not limited to, vascular permeability, inflammation, asthma, rheumatoid arthritis or osteoarthritis.
  • Immunological disorders include, and are not limited to, transplant tissue rejection, HIV-1 or immunological disorders treated or ameliorated by PKC modulation. Dermatological disorders include, and are not limited to, psoriasis, hair loss or baldness. Oncological disorders include, and are not limited to, cancer or tumor growth (such as breast, brain, kidney, bladder, ovarian or colon cancer or leukemia) and other diseases associated with uncontrolled cell proliferation such as recurring benign tumors as well as including proliferative angiopathy and angiogenesis; and, includes use for compounds of claims 1 to 5 as an adjunct to chemotherapy and radiation therapy.
  • cancer or tumor growth such as breast, brain, kidney, bladder, ovarian or colon cancer or leukemia
  • other diseases associated with uncontrolled cell proliferation such as recurring benign tumors as well as including proliferative angiopathy and angiogenesis
  • CNS disorders include, and are not limited to, chronic pain, neuropathic pain, epilepsy, chronic neurodegenerative conditions (such as dementia or Alzheimer's disease), mood disorders (such as schizophrenia), manic depression or neurotraumatic, cognitive decline and ischemia-related diseases (as a result of head trauma (from acute ischemic stroke, injury or surgery) or transient ischemic stroke (from coronary bypass surgery or other transient ischemic conditions)).
  • compositions contemplated within this invention can be prepared according to conventional pharmaceutical techniques.
  • a pharmaceutically acceptable carrier may be used in the composition of the invention.
  • the composition may take a wide variety of forms depending on the form of preparation desired for administration including, but not limited to, intravenous (both bolus and infusion), oral, nasal, transdermal, topical with or without occlusion, intraperitoneal, subcutaneous, intramuscular or parenteral, all using forms well known to those of ordinary skill in the pharmaceutical arts.
  • one or more of the usual pharmaceutical carriers may be employed, such as water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, syrup and the like in the case of oral liquid preparations (for example, suspensions, elixirs and solutions), or carriers such as starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations (for example, powders, capsules and tablets).
  • oral liquid preparations for example, suspensions, elixirs and solutions
  • carriers such as starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations (for example, powders, capsules and tablets).
  • the compounds may alternatively be administered parenterally via injection of a formulation consisting of the active ingredient dissolved in an inert liquid carrier.
  • the injectable formulation can include the active ingredient mixed with an appropriate inert liquid carrier.
  • Acceptable liquid carriers include vegetable oils such as peanut oil, cotton seed oil, sesame oil, and the like, as well as organic solvents such as solketal, glycerol, formal, and the like.
  • aqueous parenteral formulations may also be used.
  • acceptable aqueous solvents include water, Ringer's solution and an isotonic aqueous saline solution.
  • a sterile non-volatile oil can usually be employed as solvent or suspending agent in the aqueous formulation.
  • the formulations are prepared by dissolving or suspending the active ingredient in the liquid carrier such that the final formulation contains from 0.005 to 10% by weight of the active ingredient.
  • Other additives including a preservative, an isotonizer, a solubilizer, a stabilizer and a pain-soothing agent may adequately be employed.
  • compounds of the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in that art.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • tablets and capsules represent an advantageous oral dosage unit form, wherein solid pharmaceutical carriers are employed. If desired, tablets may be sugar-coated or enteric-coated by standard techniques.
  • the active drug component can be combined in suitably flavored suspending or dispersing agents such as the synthetic and natural gums, including for example, tragacanth, acacia, methyl-cellulose and the like.
  • suspending or dispersing agents such as the synthetic and natural gums, including for example, tragacanth, acacia, methyl-cellulose and the like.
  • Other dispersing agents include glycerin and the like.
  • the compounds of the present invention can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • liposome delivery systems such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • Liposomes containing delivery systems as well known in the art are formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
  • the instant pharmaceutical composition will generally contain a per dosage unit (e.g., tablet, capsule, powder, injection, teaspoonful and the like) from about 0.001 to about 100 mg/kg.
  • the instant pharmaceutical composition contains a per dosage unit of from about 0.01 to about 50 mg/kg of compound, and preferably from about 0.05 to about 20 mg/kg.
  • Methods are known in the art for determining therapeutically effective doses for the instant pharmaceutical composition.
  • the therapeutically effective amount for administering the pharmaceutical composition to a human for example, can be determined mathematically from the results of animal studies.
  • a wavy line indicates bond attachment to a larger structure that is not shown but is otherwise identical to the larger compound of which the compound fragment is drawn.
  • Representative compounds of the present invention can be synthesized in accordance with the general synthetic methods described below and are illustrated more particularly in the schemes that follow. Since the schemes are illustrations, the invention should not be construed as being limited by the chemical reactions and conditions expressed. The preparation of the various starting materials used in the schemes is well within the skill of persons versed in the art.
  • Compound A1 (wherein A is selected from nitrogen and E is selected from carbon for compounds of Formula (Ia) and A and E are selected from nitrogen for compounds of Formula (Ic)) was dissolved in a suitable solvent and then cooled. Trimethyltin chloride was added under an inert atmosphere to react with Compound A1 (below) and then BuLi was added. The reaction was washed with an aqueous solvent and the product Compound A2 was purified. Compound A2 was reacted with a 2,3-dichloromaleimide Compound A3 in the presence of PdCl 2 (PPh 3 ) 2 and LiCl in a suitable solvent. The product Compound A4 may then be purified by column chromatography.
  • Chloro-indoylmaleimide Compound A2 (wherein A is selected from nitrogen and E is selected from carbon for compounds of Formula (Ig) and A and E are selected from nitrogen for compounds of Formula (Ih)) and Compound B1 were diluted in a suitable solvent and reacted in the presence of LiCl and dichlorobis(triphenylphosphine)palladium(II) in an inert atmosphere.
  • the Compound A2 protecting group was removed from an intermediate of Compound B1 by reaction with TFA in a suitable solvent to yield the product Compound B2 .
  • a hydroxy polyalkoxy chain Compound C1 may be reacted with TsCl or MsCl to produce a polyalkoxy chain Compound C2 or Compound C3, respectively (prepared as described in Bender, S. L. and Gauthier, D. R., Tetrahedron Lett., 1996, 37(1), 13-16).
  • the Compound A4 (wherein A and E are independently selected from the group consisting of a carbon atom and a nitrogen atom) was dissolved in a suitable solvent with Cs 2 CO 3 at an elevated temperature.
  • the polyalkoxy chain Compound C2 or Compound C3 was dissolved in a suitable solvent and was added slowly to the reaction mixture. The reaction was then extracted and purified to yield the product Compound C4.
  • T f O CF 3 SO 3
  • T s O toluleneSO 3
  • the Compound C4 was dissolved in an alcohol, then a base and heated to reflux. The reaction was acidified to form a precipitated Compound C5.
  • Compound C5 was dissolved in a suitable solvent containing HMDS and heated for a time and at a temperature sufficient to produce Compound C6. The product Compound C6 may then be purified by column chromatography.
  • the Compound A4 (wherein A and E are independently selected from the group consisting of a carbon atom and a nitrogen atom) was diluted in a suitable solvent containing Cs 2 CO 3 and reacted at an elevated temperature with Compound D1 (dibromo(CH 2 ) 1-4 alkyl; wherein X is a carbon or a nitrogen atom).
  • Compound D1 dibromo(CH 2 ) 1-4 alkyl; wherein X is a carbon or a nitrogen atom.
  • elevated temperature is used herein to refer to temperatures that are preferably greater than 22° C and preferably below the reflux temperature. It is understood that those in the art will be able to vary the time and temperature of these reactions to optimize product production.
  • the product was extracted and purified to yield Compound D2.
  • the product Compound D2 was dissolved in an alcohol and base and was heated to reflux. Then the reaction was acidified to form a precipitated intermediate which was dissolved in a suitable solvent containing HMDS and was heated.
  • the product Compound D3 was purified by column chromatography.
  • the Compound A4 (wherein A and E are independently selected from the group consisting of a carbon atom and a nitrogen atom) was diluted in a suitable solvent containing Cs 2 CO 3 and reacted at elevated temperature with a Compound E1 (wherein a is (CH 2 ) 1-6 alkyl). The product was extracted and purified to yield a Compound E2.
  • the Compound E2 was reacted with R 6 NH 2 in the presence of DIEA (N,N-diisopropylethylamine) in THF at an elevated temperature, then cooled and evaporated to give a Compound E3.
  • the Compound E3 was dissolved in an alcohol and base and heated to reflux. The reaction was then acidified and evaporated. The resulting solid was treated with ammonium acetate at elevated temperatures, cooled, and extracted to provide Compound E4 .
  • the Compound A4 (wherein A and E are independently selected from the group consisting of a carbon atom and a nitrogen atom) was diluted in a suitable solvent containing Cs 2 CO 3 and reacted at elevated temperature with a Compound F1 (dihalo(CH 2 ) 1-6 alkyl). The product was extracted and purified to yield a Compound F2.
  • the Compound F2 was reacted with a Compound F3 NHR 6 (CH 2 ) 1-6 NR 7 (CH 2 ) 1-6 NHR 8 in the presence of DIEA (N,N-diisopropylethylamine) and KI in THF at an elevated temperature.
  • the product was cooled and evaporated to give a Compound F4.
  • the Compound F4 was dissolved in an alcohol and base and heated to reflux. The reaction was then acidified and evaporated. The resulting solid was treated with ammonium acetate at elevated temperatures, cooled and extracted to form Compound F5
  • the Compound F2 was reacted with a Compound F6 NHR 6 (CH 2 ) 1-6 NHR 7 or Compound F8 NHR 6 to give a product Compound F7 having 2 nitrogen atoms within the macrocyclic ring or a product Compound F9 having 1 nitrogen atom within the macrocyclic ring.
  • the unsubstituted imide Compound F10 and Compound F11 may be obtained from Compound F7 and Compound F9 , respectively.
  • a mixture of Compound G1 (wherein A and E are independently selected from the group consisting of a carbon atom and a nitrogen atom) and Compound G2 (wherein b and c are independently selected from (CH 2 ) 0-5 alkyl) were dissolved in a suitable solvent and then reacted at an elevated temperature in the presence of cesium carbonate.
  • the reaction was filtered, evaporated and the residue was purified to give Compound G3 .
  • Compound G4 was dissolved in an appropriate solvent under an inert atmosphere and HOBT and DCC were added.
  • the reaction was stirred and ammonium hydroxide was slowly added and the reaction was stirred again.
  • the reaction was filtered and the filtrate was collected and extracted with an aqueous solvent.
  • the ester Compound G3 and amide Compound G7 were dissolved in a suitable solvent under an inert atmosphere and were cooled. Then 1.0 M potassium t -butoxide in THF was slowly added to the reaction mixture. The resulting mixture was stirred under cool conditions, allowed to warm and then stirred again. Then concentrated HCl was added and the reaction was stirred again. The mixture was partitioned between EtOAc and H 2 O. Two layers were separated and the aqueous layer was extracted with EtOAc. The combined extracts were washed with water, saturated aq. NaHCO 3 and brine, then dried and evaporated to give a Compound G8 . The Compound G8 was dissolved in a solvent containing pyridine and then Ms 2 O was added.
  • Trimethyl tin chloride (26.5 mL, 1 M in THF, 26.5 mmol) was added to a THF solution (15 mL) of 7-aza-1-(tert-butyloxycarbonyl)-3-iodoindole Compound 1a (1.82 g, 5.3 mmol, Kelly, T. A., J. Med Chem . 1997, 40 , 2430) at -78 °C under nitrogen.
  • n-BuLi (10 mL, 1.6 M in hexane, 16 mmol) was added dropwise at -78 °C and the reaction was allowed to warm up to 20 °C overnight. Water (4 mL) was added and the solvent was removed under vacuum.
  • Tetraethylenebismesylate Compound 1f (0.252 g, 0.72 mmol) in DMF (5.4 mL) was added via syringe pump for 3 h to a suspension of Cs 2 CO 3 (0.51 g 1.56 mmol) and starting material Compound 1d (0.162 g, 0.48 mmol) in DMF (24 mL) at 100 °C. After addition was completed the reaction mixture was cooled to 20 °C and stirred for 3 h. The reaction mixture was diluted with NH 4 Cl (aq) and the product was extracted into CH 2 Cl 2 . The organic layer was washed with water, dried (Na 2 SO 4 ) and concentrated.
  • Pentaethylenebismesylate Compound 1g (0.3 g, 0.76 mmol) in DMF (6 mL) was added via syringe pump for 4 h to a suspension of Cs 2 CO 3 (0.41 g, 1.27 mmol) and starting material Compound 1d (0.2 g, 0.58 mmol) in DMF (18 mL) at 100 °C. After addition was completed, the reaction mixture was cooled to 20 °C and stirred for 3 h. The reaction mixture was diluted with NH 4 Cl (aq) after it was cooled to 0 °C in an ice bath. The product was extracted into CH 2 Cl 2 . The organic layer was washed with water, dried (Na 2 SO 4 ) and concentrated.
  • Triethylenebismesylate Compound 1e (0.58 g, 1.9 mmol) in DMF (15 mL) was delivered via syringe pump for 3 hours to a suspension of Cs 2 CO 3 (1.0 g, 3.2 mmol) and starting material Compound 2a (0.5 g, 1.5 mmol, prepared as described in Synthesis , 1995 , 1511) in DMF (40 mL) at 100 °C. Next the reaction mixture was cooled to 20 °C and stirred for 3 h. The reaction mixture was diluted with NH 4 Cl (aq) and the product was extracted into CH 2 Cl 2 . The organic layer was washed with water, dried (Na 2 SO 4 ) and concentrated.
  • Triethylenebismesylate Compound 1f (1.9 mmol) in DMF (15 mL) was delivered via syringe pump for 3 hours to a suspension of Cs 2 CO 3 (1.0 g, 3.2 mmol) and starting material Compound 2a (0.5 g, 1.5 mmol) in DMF (40 mL) at 100 °C.
  • the reaction mixture was cooled to 20 °C and stirred for 2 h.
  • the reaction mixture was diluted with NH 4 Cl (aq) and the product was extracted into CH 2 Cl 2 .
  • the organic layer was washed with water, dried (Na 2 SO 4 ) and concentrated.
  • Pentaethylenebismesylate Compound 1g (0.75 g, 1.9 mmol) in DMF (15 mL) was added via syringe pump overnight to a suspension of Cs 2 CO 3 (1.0 g, 3.2 mmol) and starting material Compound 2a (0.5 g ,1.5 mmol) in DMF (40 mL) at 100 °C. The reaction mixture was cooled to 20 °C and stirred for 2 h. The reaction mixture was diluted with NH 4 Cl (aq) and the product was extracted into CH 2 Cl 2 . The organic layer was washed with water, dried (Na 2 SO 4 ) and concentrated.
  • a dihalo substituted aryl/heteroaryl Compound 3d (such as ⁇ , ⁇ '-dibromo- m -xylene; wherein X is a carbon atom and halo is a bromo atom) (200 mg, 0.756 mmol) in DMF (10 mL) was added over a 2 h period with a syringe pump to a slurry of Compound 3c (246 mg, 0.72 mmol) and Cs 2 CO 3 (394 mg, 1.2 mmol) in DMF (20 mL) at 100 °C was held at 100 °C for 20 h. The mixture was concentrated under vacuum.
  • a dihalo substituted aryl/heteroaryl Compound 3d (such as 2,6-bis(chloromethyl)pyridine; wherein X is a nitrogen atom and halo is a chloro atom) (133 mg, 0.756 mmol) in DMF (20 mL) was added over a 2 h period with a syringe pump to a slurry of Compound 3c (246 mg, 0.72 mmol) and Cs 2 CO 3 (394 mg, 1.2 mmol) in DMF (20 mL) at 100 °C and was held at 100 °C for 20 h. The reaction mixture was concentrated under vacuum. Water was added and the residue was extracted with ethyl acetate and then with CH 2 Cl 2 .
  • the crude product was chromatographed (silica gel, DCM/MeOH/NH 4 OH, from 95:3:2 to 93:5:2) to produce the target Compound 12 (38.5 mg).
  • the DCM solution was extracted four times with 5% NaHCO 3 (150 mL); the combined aqueous solution was treated with sodium chloride (190 g) and extracted with ethyl acetate (300 mL) six times. The organic extract was dried (Na 2 SO 4 ) and evaporated in vacuo to a solid, which was triturated with diethyl ether (100 mL) and filtered to afford a white solid Compound 7d (3.52 g, 67%).
  • Triethylamine (0.47 mL, 3.35 mmol) and MsCl (0.13 mL, 1.67 mmol) were added to a solution of the diol Compound 8a (87 mg, 0.167 mmol) in CH 2 Cl 2 (1.5 mL) at 0 °C. After stirring at 20 °C for 15 min, the mixture was quenched with water (0.5 mL) and then diluted with CH 2 Cl 2 (5 mL). After the layers were separated, the aqueous phase was extracted with CH 2 Cl 2 (3 x 5 mL) and the organic layers were combined, dried (Na 2 SO 4 ) and concentrated.
  • the 2-bromoethylether Compound 5f (0.2 mL, 1.57 mmol) was added to a mixture of Compound 1d (54 mg, 0.16 mmol), Cs 2 CO 3 (205 mg, 0.63 mmol) and DMF (5.0 mL). After heating at 40 °C for 1.5 h, the mixture was stirred at 20 °C for 12 h, then filtered through Celite and diluted with EtOAc. The organic layer was washed with water (3 x 5 mL), dried (Na 2 SO 4 ) and concentrated.
  • the mixture was cooled to 20 °C, diluted with water (3.0 mL), made basic with 20% aqueous NaOH to achieve a pH of 10 and extracted with EtOAc (3 x 25 mL). The combined organic layers were dried (Na 2 SO 4 ) and concentrated.
  • Methanesulfonic acid (0.5 mL) was added to a solution of the Compound 121 (5 mg, 0.008 mmol) in CH 2 Cl 2 (1.0 mL). The mixture was stirred at 20 °C for 6 h, then ammonium hydroxide was carefully added to make the mixture basic. The mixture was extracted with EtOAc (2 x 10 mL) and the organic layers were combined, washed with water (5 mL) and brine (5 mL), then dried (Na 2 SO 4 ) and concentrated.
  • Methanesulfonic acid (0.2 mL) was added to a solution of Compound 13a (6 mg, 0.009 mmol) in CH 2 Cl 2 (1.0 mL). After the mixture was stirred at 20 °C for 2 h, ammonium hydroxide was carefully added to make the mixture basic. The mixture was then extracted with EtOAc (2 x 10 mL) and the organic layers were combined, washed with water (5 mL) and brine (5 mL), then dried (Na 2 SO 4 ) and concentrated.
  • Triethylamine (0.95 mL, 6.84 mmol) at 0 °C was added to a solution of the diol Compound 14d (254 mg, 1.14 mmol) in CH 2 Cl 2 (4 mL), followed by MsCl (0.35 mL, 4.56 mmol). The mixture was stirred at 20 °C for 1.5 h, diluted with diethyl ether (20 mL) and washed with 5% HCl (5 mL). The layers were separated and the organic phase was discarded. The aqueous phase was diluted with CH 2 Cl 2 (10 mL) and made basic with 5% NaOH (5 mL).
  • Triethylamine (0.41 mL, 2.97 mmol) and Mscl (0.23 mL, 2.97 mmol) at 0 °C were added to a methylene chloride (2.5 mL) solution of Compound 15e (120 mg, 0.59 mmol). The mixture was stirred at 20 °C for 2 h and quenched with water. The layers were separated and the aqueous phase was extracted with CH 2 Cl 2 (2 x 20 mL).
  • the crude Compound 16f was mixed with methylene chloride (1 mL), then methanesulfonic acid (0.3 mL) was added. The mixture was stirred at 20 °C for several hours until Compound 16f was no longer detected by MS. The mixture was cooled in an ice bath, carefully quenched with ammonium hydroxide and extracted with EtOAc (3 x 15 mL). The extracts were washed with water (10 mL) and brine (10 mL), then dried (Na 2 SO 4 ) and concentrated.
  • the compounds of the present invention were tested for biological activity in the following in-vitro and in-vivo methods.
  • PLC Protein Kinase C
  • the different human PKC isozymes (were obtained from Pan Vera, Madison WI and had been prepared as recombinant enzymes produced from a baculovirus expression vector) were added to a reaction mixture containing a test compound, 20 mM HEPES (pH 7:4), 100 ⁇ M CaCl 2 , 10 mM MgCl 2 , 100 ⁇ g/mL phosphatidylserine, 20 ⁇ g/mL diacylglycerol, 1 ⁇ M ATP, 0.8 ⁇ Ci ( 33 P)ATP, and 5 ⁇ g/mL biotinylated substrate peptide (Jing Zhao et al., J. Bio. Chem ., 1998, 273, 23072).
  • reaction was incubated for 15 min at 30°C. Reactions were terminated by the addition of streptavidin-coated SPA beads (Amersham) in a solution containing 1mM EGTA, 10mM EDTA and 100 ⁇ M ATP. Beads were allowed to settle overnight and the plates read in a Wallac MICROBETA scintillation counter (PerkinElmer Life sciences, Wellesley, MA).
  • the inhibitory activity of a compound against Glycogen Synthase Kinase 3- ⁇ (GSK 3- ⁇ ) activity was assessed using a recombinant rabbit GSK 3- ⁇ according to the procedure below.
  • the test compound was added to a reaction mixture containing Protein phosphatase inhibitor-2 (PPI-2) (Calbiochem, San Diego CA) (45 ng), rabbit GSK-3- ⁇ (New England Biolabs, Beverly MA) (0.75 units) and 33 P-ATP (1uCi) in 50 mM Tris-HCl (pH 8.0), 10 mM MgCl 2 , 0.1% BSA, 1 mM DTT, and 100 uM Sodium Vanadate. The mixture was reacted for 90 minutes at 30°C to allow phosphorylation of the PPI-2 protein and then the protein in the reaction was precipitated using 10 % trichloroacetic acid (TCA).
  • PPI-2 Protein phosphatase inhibitor-2
  • TAA 10 % trichloroacetic acid
  • the precipitated protein was collected on filter plates (MultiScreen-DV, Millipore, Bedford MA), which were subsequently washed. Finally, the radioactivity was quantified using a TopCount Scintillation Counter (Packard, Meridian CT). GSK-3 inhibitory compounds resulted in less phosphorylated PPI-2 and thus a lower radioactive signal in the precipitated protein. Staurosporine or Valproate (both available from several commercial sources), known inhibitors of GSK-3- ⁇ , were used as a positive control for screening.
  • Biotinylated peptide substrates were selected from the literature as appropriate for the enzyme being evaluated.
  • Assay conditions vary slightly for each protein kinase, for example, insulin receptor kinase requires 10 mM MnCl 2 for activity and Calmodulin-dependent protein kinase requires calmodulin and 10 mM CaCl 2 .
  • the reaction mixture was dispensed into the wells of a streptavidin coated Flashplate and 1 ⁇ L drug stock in 100% DMSO was added to a 100 ⁇ L reaction volume resulting in a final concentration of 1% DMSO in the reaction.
  • the reaction was incubated for one hour at 30°C in the presence of compound. After one hour the reaction mix was aspirated from the plate and the plate was washed with PBS containing 100 mM EDTA. The plate was read on a scintillation counter to determine 33 P- ⁇ -ATP incorporated into the immobilized peptide.
  • Test compounds were assayed in duplicate at 8 concentrations (100 uM, 10 uM, 1 uM, 100 nM, 10 nM, 1 nM, 100 pM, 10 pM). A maximum and minimum signal for the assay was determined on each plate.
  • VEGF-R vascular endothelial growth factor receptor-2
  • vascular endothelial growth factor receptor-2 vascular endothelial growth factor receptor-2
  • Protein Kinase A is the catalytic subunit of cAMP dependent protein kinase-A purified from bovine heart (Upstate Biotech, Lake Placid, NY, Cat#14-114).
  • CDK1 cyclin dependent kinase 1
  • CDK1 cyclin dependent kinase 1
  • Casein Kinase-1 is a protein truncation at amino acid 318 of the C-terminal portion of the rat CKI delta isoform produced in E.coli (New England Biolabs, Beverly, MA, Cat. #6030).
  • Insulin Receptor Kinase consists of residues 941-1313 of the cytoplasmic domain of the beta-subunit of the human insulin receptor (BIOMOL, Madison Meeting, PA, Cat. #SE-195).
  • Calmodulin Kinase (calmodulin-dependent protein kinase 2) is a truncated version of the alpha subunit of the rat protein produced in insect cells (New England Biolabs, Beverly, MA, Cat. #6060).
  • MAP Kinase is the rat ERK-2 isoform containing a polyhistidine tag at the N-terminus produced in E.coli and activated by phosphorylation with MEK1 prior to purification (BIOMOL, Madison Meeting, PA, Cat. #SE-137).
  • EGFR epimal growth factor receptor
  • IC 50 data for certain compounds of the invention tested against various kinases are shown in Table 2. For compounds where a kinase IC 50 value is >10, there was no observed 50% inhibition at the highest dose tested for that kinase nor was an inhibition maxima observed.
  • Glycogen content of L6 muscle cells was measured according to the method described in Berger and Hayes, Anal. Biochem., 1998, 261, 159-163.
  • L6 cells were serum starved overnight in alpha-MEM containing 0.1%.
  • cells were washed three times with 300 ⁇ L KRPH buffer (150 mM NaCl, 5 mM KCl, 2.9 mM Na 2 HPO 4 , 1.25 mM MgSO 4 , 1.2 mM CaCl 2 , 10 mM HEPES, pH 7.4) and labeled with 200 ⁇ L alpha-MEM containing 5.5 mM 14 C-Glucose (0.1 ⁇ Ci) in the presence of vehicle (DMSO) or compounds.
  • DMSO vehicle
  • cells were washed three times with ice-cold PBS and glycogen was precipitated for 2 hours using ice-cold 66% EtOH. Precipitated glycogen was then washed three times with ice-cold 66% EtOH and 14 C-glycogen was quantified using a TopCount (Packard).

Landscapes

  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Cardiology (AREA)
  • Pain & Pain Management (AREA)
  • Diabetes (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Dermatology (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • Pulmonology (AREA)
  • Virology (AREA)
  • Urology & Nephrology (AREA)
  • Rheumatology (AREA)
  • Hematology (AREA)
  • Communicable Diseases (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Endocrinology (AREA)
  • Vascular Medicine (AREA)
  • Emergency Medicine (AREA)
  • Transplantation (AREA)
  • Oncology (AREA)
  • Obesity (AREA)

Claims (24)

  1. Verbindung der Formel (Ia1):
    Figure 01070001
    Worin R4, R2, und R5 abhängig voneinander ausgewählt sind aus: R 4 R 2 R 5 -(CH2)2- -O-(CH2)2-O- -(CH2)2-; -(CH2)2- -O-(CH2)2-O-(CH2)2-O- -(CH2)2-; -(CH2)2- -O-(CH2)2-O-(CH2)2-O-(CH2)2-O- -(CH2)2-; -(CH2)2- -O-(CH2)2-O-(CH2)2-O-(CH2)2-O-(CH2)2-O- -(CH2)2-; -(CH2)2- -O-(CH2)2-N(Et)-(CH2)2-O- -(CH2)2-; -(CH2)2- -O-(CH2)2-N(Me)-(CH2)2-O- -(CH2)2-; -(CH2)2- -O-(CH2)2-N(i-Pr)-(CH2)2-O- -(CH2)2-; -(CH2)2- -N(Me)-(CH2)2-N(Me)-(CH2)2-N(Me)- -(CH2)2-; -(CH2)2- -O-(CH2)2-N(2-hydroxy-Et)-(CH2)2-O- -(CH2)2-; und -(CH2)2- -O-(CH2)2-O-(CH2)2-N(Me)- -(CH2)3-.
    und pharmazeutisch akzeptable Salze davon.
  2. Verbindung der Formel (Ib1):
    Figure 01080001
       worin R4, R2, und R5 abhängig voneinander ausgewählt sind aus: R 4 R 2 R 5 -(CH2)2- -O-(CH2)2-O-(CH2)2-O- -(CH2)2-; -(CH2)2- -O-(CH2)2-O-(CH2)2-O-(CH2)2-O- -(CH2)2-; -(CH2)2- -O-(CH2)2-O-(CH2)2-O-(CH2)2-O-(CH2)2-O- -(CH2)2-; -(CH2)2- -O-(CH2)2-N(Et)-(CH2)2-O- -(CH2)2-; -(CH2)2- -O-(CH2)2-S-(CH2)2-O- -(CH2)2-; -(CH2)5- -NH- -(CH2)5-; -(CH2)5- -N(Et)- -(CH2)5-; -(CH2)5- -NH- -(CH2)4-; -(CH2)5- -N(Et)- -(CH2)4-; -(CH2)4- -2,6-pyridinyl- -(CH2)4-; -(CH2)4- -C(O)-(CH2)2- -(CH2)4-; -(CH2)4- -C(O)- -(CH2)4-; -CH2- -CH[R](OH)-(CH2)6-CH[R](OH)- -CH2-; und -(CH2)2- -O-(CH2)2-O- -(CH2)2-.
    und pharmazeutisch akzeptable Salze davon.
  3. Verbindung der Formel (If1):
    Figure 01080002
       worin R4, R2, und R5 abhängig voneinander ausgewählt sind aus: R 4 R 2 R 5 -(CH2)2- -O-(CH2)2-N(Me)-(CH2)2-O- -(CH2)2-; -(CH2)2- -O-(CH2)2-N(Et)-(CH2)2-O- -(CH2)2-; und -(CH2)2- -O-(CH2)2-N(2-OMe-Et)-(CH2)2-O- -(CH2)2-.
    und pharmazeutisch akzeptable Salze davon.
  4. Verbindung der Formel (Ii1):
    Figure 01090001
       worin R4, R2, und R5 abhängig voneinander ausgewählt sind aus: R 4 R 2 R 5 -CH2- -1,3-phenyl- -CH2-; und -CH2- -2,6-pyridinyl- -CH2-.
    und pharmazeutisch akzeptable Salze davon.
  5. Verbindung der Formel (Ij1):
    Figure 01090002
    worin R4, R2, und R5 abhängig voneinander ausgewählt sind aus: R 4 R 2 R 5 -(CH2)2- -O-(CH2)2-O- -(CH2)2- und -(CH2)2- -O-(CH2)2-O-(CH2)2-O- -(CH2)2-
    und pharmazeutisch akzeptable Salze davon.
  6. Pharmazeutische Zusammensetzung, umfassend eine Verbindung nach einem der Ansprüche 1 bis 5 und einen pharmazeutisch akzeptablen Träger.
  7. Pharmazeutische Zusammensetzung, hergestellt durch Mischen einer Verbindung nach einem der Ansprüche 1 bis 5 und einem pharmazeutisch akzeptablen Träger.
  8. Verfahren zur Herstellung einer pharmazeutischen Zusammensetzung, umfassend Mischen einer Verbindung nach einem der Ansprüche 1 bis 5 mit einem pharmazeutisch akzeptablen Träger.
  9. Verwendung einer Verbindung nach einem der Ansprüche 1 bis 5 zur Herstellung eines Medikaments zur Verwendung bei der Behandlung einer Kinase-vermittelten Erkrankung.
  10. Verwendung nach Anspruch 9, wobei die Kinase-vermittelte Erkrankung mit der Aktivität von Protein-Kinase C und Glykogensynthase Kinase-3-Aktivität assoziiert ist.
  11. Verwendung nach Anspruch 10, wobei die Kinase ausgewählt ist aus der Gruppe bestehend aus Protein-Kinase C α, Protein-Kinase C β-II, Protein-Kinase C γ und Glykogensynthase Kinase-3 β.
  12. Verwendung nach Anspruch 9, wobei die Kinase-vermittelte Erkrankung eine Dual-Kinase-vermittelte Erkrankung ist und wobei mindestens eine der Kinasen ausgewählt ist aus der Gruppe bestehend aus Protein-Kinase C und Glykogensynthase Kinase-3.
  13. Verwendung nach Anspruch 10, wobei mindestens eine Kinase ausgewählt ist aus der Gruppe bestehend aus Protein-Kinase C α, Protein-Kinase C β-II, Protein-Kinase C γ und Glykogensynthase Kinase-3 β.
  14. Verwendung nach Anspruch 9, wobei die Kinase-vermittelte Erkrankung ausgewählt ist aus der Gruppe bestehend aus kardiovaskulären Erkrankungen, Diabetes, Diabetes-assoziierten Erkrankungen, entzündlichen Erkrankungen, immunologischen Erkrankungen, dermatologischen Erkrankungen, onkologischen Erkrankungen und CNS-Erkrankungen.
  15. Verwendung nach Anspruch 14, wobei die kardiovaskulären Erkrankungen ausgewählt sind aus der Gruppe bestehend aus akutem Schlaganfall, Herzversagen, kardiovaskulärer Ischämie, Thrombose, Arteriosklerose, Bluthochdruck, Restenose, Retinopathie oder Frühreife- und Alters-zusammenhängende makularer Degeneration.
  16. Verwendung nach Anspruch 14, wobei die Diabetes ausgewählt ist aus der Gruppe bestehend aus Insulin-abhängiger Diabetes und Typ II nicht-Insulin-abhängiger Diabetes Mellitus.
  17. Verwendung nach Anspruch 14, wobei die Diabetes-assoziierten Erkrankungen ausgewählt sind aus der Gruppe bestehend aus beeinträchtigter Glukosetoleranz, diabetischer Retinopathie, proliferativer Retinopathie, retinalem Venenverschluß, makularen Ödemen, Kardiomyopathie, Nephropathie und Neuropathie.
  18. Verwendung nach Anspruch 14, wobei die entzündlichen Erkrankungen ausgewählt sind aus der Gruppe bestehend aus vaskulärer Permeabilität, Entzündung, Asthma, rheumatoider Arthritis und Osteoarthritis.
  19. Verwendung nach Anspruch 14, wobei die immunologischen Erkrankungen ausgewählt sind aus der Gruppe bestehend aus Transplantatgewebe-Abstoßung, HIV-1 und PCK-vermittelten immunologischen Erkrankungen.
  20. Verwendung nach Anspruch 14, wobei die dermatologischen Erkrankungen ausgewählt sind aus der Gruppe bestehend aus Psoriasis, Haarverlust und Glatzköpfigkeit.
  21. Verwendung nach Anspruch 14, wobei die onkologischen Erkrankungen ausgewählt sind aus der Gruppe bestehend aus Krebs, Tumorwachstum, unkontrollierter Zellproliferation, proliferativer Angiopathie und Angiogenese.
  22. Verwendung nach Anspruch 14, wobei die Erkrankungen des zentralen Nervensystems ausgewählt sind aus der Gruppe bestehend aus chronischen Schmerzen, neuropathischen Schmerzen, Epilepsie, chronischen neurodegenerativen Zuständen, Demenz, Alzheimererkrankung, Stimmungserkrankungen, Schizophrenie, manischer Depression und neurotraumatischem kognitivem Nachlassen und Ischämie-vermittelten Erkrankungen.
  23. Verwendung nach Anspruch 9, wobei die Behandlung die Verabreichung einer therapeutisch effektiven Menge der pharmazeutischen Zusammensetzung von 0,001 mg/kg/Tag bis 300 mg/kg/Tag umfaßt.
  24. Verwendung einer Verbindung nach einem der Ansprüche 1 bis 5 zur Herstellung eines Medikaments zur Verwendung als ein Zusatz bei der Chemotherapie und Bestrahlungstherapie.
EP01996227A 2000-12-08 2001-12-06 Makroheterocyclische verbindungen als kinase inhibitoren Expired - Lifetime EP1345946B1 (de)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US25416100P 2000-12-08 2000-12-08
US254161P 2000-12-08
PCT/US2001/047866 WO2002046197A1 (en) 2000-12-08 2001-12-06 Macroheterocylic compounds useful as kinase inhibitors

Publications (2)

Publication Number Publication Date
EP1345946A1 EP1345946A1 (de) 2003-09-24
EP1345946B1 true EP1345946B1 (de) 2005-08-10

Family

ID=22963161

Family Applications (1)

Application Number Title Priority Date Filing Date
EP01996227A Expired - Lifetime EP1345946B1 (de) 2000-12-08 2001-12-06 Makroheterocyclische verbindungen als kinase inhibitoren

Country Status (12)

Country Link
US (1) US6828327B2 (de)
EP (1) EP1345946B1 (de)
JP (1) JP2004526676A (de)
AT (1) ATE301661T1 (de)
AU (2) AU2737102A (de)
CA (1) CA2431187A1 (de)
DE (1) DE60112611T2 (de)
ES (1) ES2245994T3 (de)
HK (1) HK1055956A1 (de)
MX (1) MXPA03005139A (de)
RU (1) RU2275373C2 (de)
WO (1) WO2002046197A1 (de)

Families Citing this family (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003215325B8 (en) 2002-03-05 2008-10-09 Eli Lilly And Company Purine derivatives as kinase inhibitors
EP1487822B1 (de) 2002-03-08 2007-08-01 Eli Lilly And Company Pyrrol-2,5-dion derivate und deren verwendung als gsk-3 inhibitoren
EP1506192B1 (de) * 2002-05-08 2008-02-27 Janssen Pharmaceutica N.V. Substituierte pyrroline als kinase inhibitoren
TWI324604B (en) 2003-06-18 2010-05-11 Novartis Ag New use of staurosporine derivatives
WO2005069906A2 (en) * 2004-01-16 2005-08-04 Yale University Methods and compositions relating to vascular endothelial growth factor and th2 mediated inflammatory diseases
US20070282016A1 (en) * 2004-09-17 2007-12-06 Jenrow Kenneth A Methods and compositions for use of angiogenesis inhibitors in the prevention and/or control of epilepsy
US8017395B2 (en) 2004-12-17 2011-09-13 Lifescan, Inc. Seeding cells on porous supports
EP1888123B1 (de) 2005-06-08 2013-01-09 Janssen Biotech, Inc. Zelltherapie für augendegeneration
US20070088019A1 (en) * 2005-09-29 2007-04-19 Han-Cheng Zhang Macroheterocyclic compounds as kinase inhibitors
CA2645289A1 (en) * 2006-03-10 2007-09-20 Janssen Pharmaceutica, N.V. Pyridine-containing macroheterocylic compounds as kinase inhibitors
US8741643B2 (en) 2006-04-28 2014-06-03 Lifescan, Inc. Differentiation of pluripotent stem cells to definitive endoderm lineage
EP2094689B1 (de) 2006-12-19 2013-04-03 Novartis AG Indolylmaleimidderivate als kinaseinhibitoren
US9080145B2 (en) 2007-07-01 2015-07-14 Lifescan Corporation Single pluripotent stem cell culture
EP2185693B1 (de) 2007-07-31 2019-07-03 Lifescan, Inc. Differenzierung menschlicher embryonaler stammzellen
ATE523585T1 (de) 2007-11-27 2011-09-15 Lifescan Inc Differenzierung menschlicher embryonaler stammzellen
CA2710644C (en) 2007-12-24 2016-03-29 Tibotec Pharmaceuticals Macrocyclic indoles as hepatitis c virus inhibitors
CA2959401C (en) 2008-02-21 2021-12-07 Centocor Ortho Biotech Inc. Methods, surface modified plates and compositions for cell attachment, cultivation and detachment
US8623648B2 (en) * 2008-04-24 2014-01-07 Janssen Biotech, Inc. Treatment of pluripotent cells
CN102159703B (zh) 2008-06-30 2015-11-25 森托科尔奥索生物科技公司 多能干细胞的分化
RU2518471C2 (ru) * 2008-08-14 2014-06-10 Тиботек Фармасьютикалз Макроциклические индольные производные, применимые в качестве ингибиторов вируса гепатита с
US9012218B2 (en) 2008-10-31 2015-04-21 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
EP2346988B1 (de) 2008-10-31 2017-05-31 Janssen Biotech, Inc. Differenzierung menschlicher embryonaler stammzellen in die pankreatisch-endokrine linie
BRPI0921996A2 (pt) 2008-11-20 2015-08-18 Centocor Ortho Biotech Inc Métodos e composições para cultura e ligação de células em substratos planos.
EP3260534A1 (de) 2008-11-20 2017-12-27 Janssen Biotech, Inc. Pluripotente stammzellenkultur auf mikroträgern
SG177481A1 (en) 2009-07-20 2012-02-28 Janssen Biotech Inc Differentiation of human embryonic stem cells
KR101786735B1 (ko) 2009-07-20 2017-10-18 얀센 바이오테크 인코포레이티드 인간 배아 줄기 세포의 분화
CA2768644A1 (en) 2009-07-20 2011-01-27 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
EP2482837A4 (de) 2009-09-29 2013-05-22 Joslin Diabetes Center Inc Verwendung von proteinkinase-c-delta (pkcd-)hemmern zur behandlung von diabetes, adipositas und leberverfettung
SG181822A1 (en) 2009-12-23 2012-08-30 Centocor Ortho Biotech Inc Differentiation of human embryonic stem cells
RU2701335C2 (ru) 2009-12-23 2019-09-25 Янссен Байотек, Инк. Способ получения популяции панкреатических эндокринных клеток, соэкспрессирующих nkx6.1 и инсулин, и способ лечения диабета
EP2348360B1 (de) * 2010-01-25 2017-09-27 Rohm and Haas Electronic Materials LLC Photoresist, der eine stickstoffhaltige Verbindung umfasst
WO2011102907A1 (en) * 2010-02-22 2011-08-25 Blanchette Rockefeller Neurosciences Institute Alzheimer's disease-specific alterations of protein kinase c epsilon (pkc-epsilon) protein levels
BR112012022145A2 (pt) 2010-03-01 2020-10-06 Janssen Biotech, Inc. métodos para purificar células derivadas de células-tronco pluripotentes
KR101903562B1 (ko) 2010-05-12 2018-10-02 얀센 바이오테크 인코포레이티드 인간 배아 줄기 세포의 분화
EP2853589B1 (de) 2010-08-31 2017-12-27 Janssen Biotech, Inc. Differenzierung menschlicher embryonaler Stammzellen
ES2585028T3 (es) 2010-08-31 2016-10-03 Janssen Biotech, Inc. Diferenciación de células madre pluripotentes
KR101851956B1 (ko) 2010-08-31 2018-04-25 얀센 바이오테크 인코포레이티드 인간 배아 줄기 세포의 분화
EP3072555B1 (de) 2011-09-02 2020-03-25 The Trustees of Columbia University in the City of New York Mk2/3-hemmer zur behandlung von stoffwechselstörungen in zusammenhang mit adipositas
KR102203056B1 (ko) 2011-12-22 2021-01-14 얀센 바이오테크 인코포레이티드 인간 배아 줄기 세포의 단일 인슐린 호르몬 양성 세포로의 분화
US9434920B2 (en) 2012-03-07 2016-09-06 Janssen Biotech, Inc. Defined media for expansion and maintenance of pluripotent stem cells
CN108103005A (zh) 2012-06-08 2018-06-01 詹森生物科技公司 人胚胎干细胞向胰腺内分泌细胞的分化
CN105008518B (zh) 2012-12-31 2020-08-07 詹森生物科技公司 在空气-液体界面处培养人胚胎干细胞以用于分化成胰腺内分泌细胞
JP6529440B2 (ja) 2012-12-31 2019-06-12 ヤンセン バイオテツク,インコーポレーテツド 膵内分泌細胞への分化のためのヒト多能性細胞の懸濁及びクラスタリング
CN105073979B (zh) 2012-12-31 2020-03-06 詹森生物科技公司 使用hb9调节子使人胚胎干细胞分化为胰腺内分泌细胞的方法
US10370644B2 (en) 2012-12-31 2019-08-06 Janssen Biotech, Inc. Method for making human pluripotent suspension cultures and cells derived therefrom
MX2016015004A (es) 2014-05-16 2017-06-27 Janssen Biotech Inc Uso de moleculas pequeñas para mejorar la expresion de mafa en celulas endocrinas pancreaticas.
AU2016287568B2 (en) 2015-07-02 2020-08-20 Turning Point Therapeutics, Inc. Chiral diaryl macrocycles as modulators of protein kinases
US20180325901A1 (en) * 2015-07-21 2018-11-15 Tp Therapeutics, Inc. Chiral diaryl macrocycles and uses thereof
RU2630958C2 (ru) * 2015-12-29 2017-09-15 федеральное государственное автономное образовательное учреждение высшего образования "Московский физико-технический институт (государственный университет)" (МФТИ) Новые макроциклические соединения, содержащие природное 3,7-диазабицикло[3.3.1]нонановое ядро и способ их получения
MA45479A (fr) 2016-04-14 2019-02-20 Janssen Biotech Inc Différenciation de cellules souches pluripotentes en cellules de l'endoderme de l'intestin moyen
TWI808958B (zh) 2017-01-25 2023-07-21 美商特普醫葯公司 涉及二芳基巨環化合物之組合療法
KR102357045B1 (ko) 2017-03-31 2022-01-28 뉴로디아그노스틱스 엘엘씨 알츠하이머 질환에 대한 림프구-기반 형태계측 시험
US20200113979A1 (en) * 2017-06-12 2020-04-16 Temple University-Of The Commonwealth System Of Higher Education Protein Kinase C-delta targeted therapy for treating radiation injury
AU2018306328B2 (en) 2017-07-28 2023-03-09 Turning Point Therapeutics, Inc. Macrocyclic compounds and uses thereof
BR112020012319A2 (pt) 2017-12-19 2020-11-24 Turning Point Therapeutics, Inc. compostos macrocíclicos para tratar doença
US20220133740A1 (en) 2019-02-08 2022-05-05 Frequency Therapeutics, Inc. Valproic acid compounds and wnt agonists for treating ear disorders

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5589472A (en) * 1992-09-25 1996-12-31 Vice; Susan F. Diindolo compounds and pharmaceutical compositions containing them
JP3581731B2 (ja) * 1993-12-07 2004-10-27 イーライ・リリー・アンド・カンパニー プロテインキナーゼc阻害物質
US5624949A (en) * 1993-12-07 1997-04-29 Eli Lilly And Company Protein kinase C inhibitors
PL329681A1 (en) 1996-05-01 1999-04-12 Lilly Co Eli Halogen-substituted inhibitor of proteinous kinase c
PT1044203E (pt) * 1997-12-31 2003-07-31 Kyowa Hakko Kogyo Kk Derivados epimericos 3' do k-252a
GB9828640D0 (en) 1998-12-23 1999-02-17 Smithkline Beecham Plc Novel method and compounds

Also Published As

Publication number Publication date
MXPA03005139A (es) 2004-01-29
CA2431187A1 (en) 2002-06-13
ES2245994T3 (es) 2006-02-01
US20030078280A1 (en) 2003-04-24
JP2004526676A (ja) 2004-09-02
RU2275373C2 (ru) 2006-04-27
WO2002046197A1 (en) 2002-06-13
US6828327B2 (en) 2004-12-07
EP1345946A1 (de) 2003-09-24
DE60112611D1 (de) 2005-09-15
DE60112611T2 (de) 2006-06-14
AU2737102A (en) 2002-06-18
AU2002227371B2 (en) 2007-05-10
HK1055956A1 (en) 2004-01-30
ATE301661T1 (de) 2005-08-15

Similar Documents

Publication Publication Date Title
EP1345946B1 (de) Makroheterocyclische verbindungen als kinase inhibitoren
AU2002227371A1 (en) Macroheterocylic compounds useful as kinase inhibitors
US7705015B2 (en) Substituted pyrroline kinase inhibitors
EP1654255B1 (de) Substituierte indazolyl(indolyl)maleimid-derivate als kinase inhibitoren
US6987110B2 (en) Substituted pyrrolines as kinase inhibitors
US20090093634A1 (en) Substituted pyrroline kinase inhibitors
US20070088019A1 (en) Macroheterocyclic compounds as kinase inhibitors
EP1900738A2 (de) Substituierte Pyrrolinkinase-Hemmer

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20030616

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

RIN1 Information on inventor provided before grant (corrected)

Inventor name: SHEN, LAN

Inventor name: WHITE, KIMBERLY, B.

Inventor name: MARYANOFF, BRUCE E.

Inventor name: SHAH, CHANDRA, R.

Inventor name: MURRAY, WILLIAM.V.

Inventor name: CONOLLY, PETER.

Inventor name: ZHANG, HAN-CHENG

Inventor name: DEANGELIS, ALAN

Inventor name: PROUTY, CATHERINE

Inventor name: KUO, GEE-HONG

Inventor name: CONWAY, BRUCE.

Inventor name: DEMAREST, KEITH.

17Q First examination report despatched

Effective date: 20040621

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20050810

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20050810

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20050810

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20050810

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REF Corresponds to:

Ref document number: 60112611

Country of ref document: DE

Date of ref document: 20050915

Kind code of ref document: P

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20051110

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20051110

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20051110

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MC

Payment date: 20051128

Year of fee payment: 5

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: E. BLUM & CO. PATENTANWAELTE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20051206

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: LU

Payment date: 20051228

Year of fee payment: 5

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20060110

REG Reference to a national code

Ref country code: HK

Ref legal event code: GR

Ref document number: 1055956

Country of ref document: HK

NLV1 Nl: lapsed or annulled due to failure to fulfill the requirements of art. 29p and 29m of the patents act
REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2245994

Country of ref document: ES

Kind code of ref document: T3

ET Fr: translation filed
PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20060511

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20061231

REG Reference to a national code

Ref country code: CH

Ref legal event code: PFA

Owner name: ORTHO-MCNEIL PHARMACEUTICAL, INC.

Free format text: ORTHO-MCNEIL PHARMACEUTICAL, INC.#U.S. ROUTE NO.202, P.O. BOX 300#RARITAN, NJ 08869-0602 (US) -TRANSFER TO- ORTHO-MCNEIL PHARMACEUTICAL, INC.#U.S. ROUTE NO.202, P.O. BOX 300#RARITAN, NJ 08869-0602 (US)

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20061206

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 16

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 17

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20171113

Year of fee payment: 17

Ref country code: DE

Payment date: 20171129

Year of fee payment: 17

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20171212

Year of fee payment: 17

Ref country code: GB

Payment date: 20171206

Year of fee payment: 17

Ref country code: BE

Payment date: 20171113

Year of fee payment: 17

Ref country code: IE

Payment date: 20171211

Year of fee payment: 17

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20180102

Year of fee payment: 17

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20171221

Year of fee payment: 17

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 60112611

Country of ref document: DE

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20181206

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20181231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190702

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181206

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181231

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181206

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181231

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181231

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181206

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20200131

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181207